Sunteți pe pagina 1din 9

Brain, Behavior, and Immunity 30 (2013) S1S9

Contents lists available at SciVerse ScienceDirect

Brain, Behavior, and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Brief Commentary

Psychoneuroimmunology and cancer: A decade of discovery, paradigm shifts,


and methodological innovations
Paige Green McDonald a,, Mary OConnell b, Susan K. Lutgendorf c,d,e,f
a
Basic Biobehavioral and Psychological Sciences Branch, Behavioral Research Program (BRP), Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute
(NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), United States
b
BRP, DCCPS, NCI, NIH, DHHS, United States
c
Department of Psychology, University of Iowa, United States
d
Department of Urology, University of Iowa, United States
e
Department of Obstetrics and Gynecology, University of Iowa, United States
f
Holden Comprehensive Cancer Center, University of Iowa, United States

a r t i c l e i n f o a b s t r a c t

Article history: This article introduces the supplement Advances in Cancer and Brain, Behavior, and Immunity and outlines
Available online 16 January 2013 important discoveries, paradigm shifts, and methodological innovations that have emerged in the past
decade to advance mechanistic and translational understanding of biobehavioral inuences on tumor
Keywords: biology, cancer treatment-related sequelae, and cancer outcomes. We offer a heuristic framework for
Biobehavioral research on biobehavioral pathways in cancer. The shifting survivorship landscape is highlighted, and
Psycho-oncology we propose that the changing demographics suggest prudent adoption of a life course perspective of can-
Tumor biology
cer and cancer survivorship. We note opportunities for psychoneuroimmunology (PNI) research to ame-
Cancer treatment
Cancer outcomes
liorate the long-term, unintended consequences of aggressive curative intent and call attention to the
critical role of reciprocal translational pathways between animal and human studies. Lastly, we briey
summarize the articles included in this compilation and offer our perspectives on future research
directions.
Published by Elsevier Inc.

1. Introduction conducted in a cancer context. In the decade prior to the NCI com-
missioned supplement, Brain, Behavior, and Immunity published only
In 2002, the National Cancer Institute (NCI), in collaboration 12 cancer-relevant articles. Since the 2003 supplement, the journal
with other Institutes and Centers of the National Institutes of has featured 128 cancer-relevant papers that have generated 3361
Health, convened a meeting of scientic experts to discuss seminal citations (data from SCOPUS, retrieved November 1, 2012), relative
research on behavioral, neural, endocrine, and immune system to 55 papers on PNI and cancer, published in other peer review jour-
interactions in health and disease. To inform the development of nals during the same time period. These bibliometric data highlight
a biobehavioral research agenda in cancer control, knowledge Brain, Behavior, and Immunity as a leading scholarly outlet for re-
was extracted from contemporary studies of neuroimmune mech- search on the biology of psychological and social experiences and
anisms of subjective experiences (e.g., stress, loneliness, and pain), the integrated mechanisms associated with cancer as a complex dis-
biological processes (e.g., circadian rhythmicity, sleep, wound heal- ease process. The current volume celebrates the 10-year anniversary
ing, sickness behavior, and apoptosis), and disease outcomes (e.g., of the 2003 supplement. This collection of invited reviews and re-
human immunodeciency virus, depression, and post-traumatic search articles captures important discoveries, paradigm shifts, and
stress disorder). Brain, Behavior, and Immunity published the Biolog- methodological innovations that have emerged in the past decade
ical Mechanisms of Psychosocial Effects on Disease supplement in to advance mechanistic and translational understanding of biobe-
February 2003. This seminal volume captured state-of-the-science havioral inuences on tumor biology, cancer treatment-related se-
reviews and commentaries by leading experts in psychoneuroim- quelae, and cancer outcomes.
munology (PNI) and served as a catalyst for biobehavioral1 research
2. Transition from cellular immunity in peripheral blood to
Corresponding author. Address: 6130 Executive Blvd, MSC 7363, Bethesda, MD tumor-relevant measurements
20892, United States. Tel.: +1 301 435 5037; fax: +1 301 435 7647.
E-mail addresses: mcdonald.paige@mail.nih.gov, pm252v@nih.gov (P. Green
McDonald).
Early clinical investigations focused almost exclusively on psy-
1
For the purpose of this introduction, we use the terms biobehavioral and chosocial modulations of the humoral and cellular immune re-
psychosocial interchangeably. sponse and, to some extent, on DNA repair (Andersen et al.,

0889-1591/$ - see front matter Published by Elsevier Inc.


http://dx.doi.org/10.1016/j.bbi.2013.01.003
S2 P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9

1994; Antoni, 2003; Kiecolt-Glaser and Glaser, 1999; Kiecolt-Gla- 4. The central nervous system as a master regulator:
ser et al., 2002). Women at an increased genetic risk for cancer implications for cancer
exhibited specic immune impairments and abnormalities in their
endocrine response to stress (Bovbjerg and Valdimarsdottir, 1993; Renewed appreciation of the landscapes that enable tumor
Dettenborn et al., 2005; Gold et al., 2003). Clinical studies docu- growth and metastatic dissemination inspire broader consider-
mented associations between depression, social support, and natu- ation of the macro-physiological milieus that potentially shape
ral killer cell activity in breast cancer patients (Levy et al., 1987; individual variability in the natural course of cancer and respon-
Levy et al., 1985; Levy et al., 1990). Other research groups observed siveness to therapies (Castano et al., 2011; Schuller and Al-Wadei,
distress/stress and social isolation-associated impairments in im- 2010). We offer the following perspective (Fig. 1). The brain, as an
mune function among breast, cervical and ovarian patients (Ander- adaptive and dynamic synthesizer of experiential and perceptual
sen et al., 1998; Antoni et al., 2009; Lutgendorf et al., 2005; Nelson processes (Ganzel et al., 2010), can participate in the complex reg-
et al., 2008; Sephton et al., 2009; Thornton et al., 2007); however, ulation of signaling systems used by the diverse array of cells and
the prognostic relevance of these associations remained uncertain structures to enable tumorigenesis. Experimental and clinical stud-
(Cohen and Rabin, 1998). Building on the clinical signicance of im- ies suggest that downstream activation of the sympathetic nervous
mune cells in ascites (Lotzova et al., 1986; Lotzova et al., 1984) and system and the hypothalamic-pituitary-adrenal axis exerts selec-
tumor-inltrating lymphocytes (Lai et al., 1996) in ovarian cancer, tive physiologic pressures that initiate molecular signaling path-
Lutgendorf and colleagues observed signicant associations be- ways involved in DNA repair, angiogenesis, cell survival,
tween psychosocial factors and the cellular immune response at inammation, invasion, metastasis, and resistance to therapy
the tumor level in a clinical sample (Lutgendorf et al., 2005). This (Antoni et al., 2006; Cole and Sood, 2012; Hara et al., 2011; Lutgen-
study, among others, signaled an important contextual transition dorf and Sood, 2011; Wu et al., 2004). Catecholamines (epineph-
for PNI studies of cancer, a transition aligned closely to advances in rine, norepinephrine, dopamine) bind to a-adrenergic receptors
cancer cell biology and emerging appreciation for target tissues (a-ARs) and b-adrenergic receptors (b-ARs), and acetylcholine
and the context in which tumors thrive (Marx, 2008). binds to families of nicotinic (nAChRs) and muscarinic (mAChRs)
receptors found on tumor cells and stromal compartments within
3. Emergence of the tumor microenvironment the microenvironment (Schuller, 2008). Neuroendocrine receptor-
mediated signaling has the documented ability to regulate leuko-
DeVita and Rosenberg (2012) recently chronicled signicant cyte gene expression, molecular processes, and functional charac-
discoveries and major events in cancer research since the founding teristics of cells within microenvironments (Badino et al., 1996;
of the New England Journal of Medicine nearly 200 years ago (DeVita Cole and Sood, 2012; Lutgendorf et al., 2003; Lutgendorf et al.,
and Rosenberg, 2012). Basic understanding of cancer biology has 2009; Schuller and Al-Wadei, 2010). Examples of observed effects
matured substantially beyond Virchows observation of the cellular include promotion of tumor cell growth, migration and invasive
origin of cancer and the view of tumors as insular masses of pro- capacity, and stimulation of angiogenesis by inducing production
liferating cancer cells (p. 646, Hanahan and Weinberg, 2011). Pro- of pro-angiogenic cytokines. Neuroendocrine hormones activate
gress has been led by milestones2 like observations from a oncogenic viruses and alter several aspects of immune function,
ploughman (Dell, 2006; Hart and Fidler, 1980; Paget, 1889), blood- including antibody production, cell trafcking, and the production
lines (Farrell, 2006; Folkman, 1971), environmental awareness and release of proinammatory cytokines (Glaser and Kiecolt-Gla-
(Schuldt, 2006), and the hallmarks of cancer (Hanahan and Wein- ser, 2005; Webster Marketon and Glaser, 2008). Although not
berg, 2000, 2011). Cancers have come to be seen as inherently com- explicitly reected in our conceptual schema, peripherally gener-
plex collections of heterogeneous pathologies that vary by tissue of ated inammatory and other innate immune mediators can signal
origin and constellation of genomic, proteomic, and metabolic alter- back into the central nervous system, stimulate afferent nerves
ations (Fidler, 2003; Hanahan and Weinberg, 2000, 2011; Vogelstein that produce local cytokines, change neuronal function, and cause
and Kinzler, 2004). Incipient mutated cells must acquire several bio- sickness behaviors as an adaptive response to systemic pressures
logical capabilities to reach full malignancy, and several environ- (Dantzer and Kelley, 2007; Dantzer et al., 2012; Dantzer et al.,
ments i.e., the primary, invasive and metastatic tumor 2008; Irwin and Cole, 2011; Kelley et al., 2003; Miller et al.,
microenvironments are created during tumorigenesis (Hanahan 2008). Immune-to-brain communication cascades are thought to
and Weinberg, 2011). In the case of solid tumors, commonly derived undergird cancer and treatment-related symptoms such as fatigue,
from epithelial cells, these microenvironments provide a safe haven depression, cognitive dysfunction, and sleep disturbance (Bower
for bidirectional communication between cancer cells and the tu- et al., 2011; Dantzer et al., 2012; Lutgendorf and Sood, 2011; Miller
mor-associated stroma. Cells that construct the microenvironments et al., 2008). Contemporary PNI remains poised to elucidate the
include pericytes, cancer-associated broblasts, endothelial cells, lo- prevalence, impact, and etiologies of cancer-related physical and
cal and bone marrow-derived stromal stem and progenitor cells, affective sequelae at different phases of cancer survival (Bower,
cancer stem cells, invasive cancer cells, and immune inammatory 2012; Dantzer et al., 2012; Haroon et al., 2012).
cells (Albini and Sporn, 2007; Joyce, 2005; Joyce and Pollard, 2009;
Langley and Fidler, 2007; McAllister and Weinberg, 2010; McCawley
and Matrisian, 2001). Inammation, a seminal biological process in 5. Shifting sands of survivorship
the onset and progression of many diseases (Haroon et al., 2012;
Nathan, 2002), has emerged as an essential enabling process for tu- Advances in prevention, detection, and treatment (DeVita and
mor growth and metastasis (Hanahan and Weinberg, 2011; Manto- Rosenberg, 2012) continue to yield signicant declines in the inci-
vani, 2009). Cytokines, chemokines, macrophages, and leukocyte dence of most cancers and death rates for all cancers combined
inltrates contribute to tumor progression by promoting invasion, (Eheman et al., 2012; Siegel et al., 2012b). These trends, combined
migration, and angiogenesis (Gonda et al., 2009; Mantovani et al., with overall increases in life expectancy, have created a booming
2008; Medrek et al., 2012; Pitroda et al., 2012; Solinas et al., [aging] cancer survivor population (p. 1996, Parry et al., 2011).
2009). Truly, it takes a village of distinct cell types and signaling sys- Siegel et al. estimated 13.7 million American cancer survivors
tems to support the tumor ecosystem. were alive in January 20123 (Siegel et al., 2012b). The majority

2 3
See http://www.nature.com/milestones/milecancer/pdf/milecancer_all.pdf. Number of survivors projected to increase to nearly 18 million by 2022.
P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9 S3

Fig. 1. Heuristic framework for research on biobehavioral risk factor inuences on clinical cancer course.

of this emergent demographic had far exceeded the 5-year survival 6. Biobehavioral risk factors in the context of cancers as chronic
benchmark. Adolescent and young adult (AYA) survivors, diagnosed diseases
at ages 15 to 29 years, have an 82% probability of survival 30 years
from diagnosis (Mertens et al., 2008). While this statistic is impres- Early prevention, detection, and treatment advances have
sive, seminal research by Oefnger, Lipshultz and others document shifted our conceptualization and management of most cancers
profound adverse long-term health-related outcomes following from acute to chronic disease models, which are often modulated
exposure to highly aggressive curative intent therapies (Lipshultz by psychosocial factors (Karelina and DeVries, 2011; Sullivan
et al., 2012; Oefnger et al., 2006). Most notably relevant to PNI, et al., 2012; Williams, 2008; Wyman et al., 2012). This paradigm
childhood cancer treatments are associated with late effects on shift further fuels our interest in psychosocial contributions to in-
the cardiovascular, central nervous, endocrine, and immune sys- tra-individual variability in cancer outcomes. Meta-analytic re-
tems. Further, survivors of adult, AYA, and pediatric cancers are views suggest stressful life experiences and depression are
at risk for recurrence and subsequent malignancies. Relative to associated with poorer survival and higher mortality across a di-
the US population, survivors experience excess morbidity and mor- verse array of cancer types (e.g., breast, lung, head and neck, hepa-
tality due to cardiac and vascular abnormalities and pulmonary tobiliary, lymphoid, and hematopoietic cancers) (Chida et al., 2008;
complications (Choi et al., 2011; Mariotto et al., 2007; Oefnger Pinquart and Duberstein, 2010; Satin et al., 2009). Prospective
and Tonorezos, 2011; Siegel et al., 2012a; Valdivieso et al., 2012). endorsement of depressive symptoms, and cortisol slope were
This landscape highlights an opportunity to use PNI paradigms to associated with decreased survival in patients with metastatic re-
understand cancer from a competing risk perspective in which nal cell carcinoma (Cohen et al., 2012). Conversely, among women
multiple factors concurrently affect risks for morbidity and mortal- with metastatic breast cancer, a decline in depressive symptoms
ity (Mell et al., 2010; Schairer et al., 2004). Although not consis- conferred survival benet (Giese-Davis et al., 2011). A recent
tently observed (Zucca et al., 2012), age at diagnosis, general life meta-analysis found the inuence of social relationships on mor-
expectancy trends, and long-term physiological sequelae of treat- tality comparable to risk conferred by tobacco and alcohol use. Fur-
ment exposure have converged to increase the prevalence of co- ther, the social relationship risk for mortality exceeded risks
morbidity or multimorbidity4 in a cancer context (Braithwaite associated with physical activity (or lack thereof) and obesity
et al., 2012; Land et al., 2012; Patnaik et al., 2011; Ritchie et al., (Holt-Lunstad et al., 2010). Inammation often mediates associa-
2011; Yood et al., 2012). tions between close relationships, depression, and chronic stress,
and health (Kiecolt-Glaser et al., 2010). Extending prior cross-sec-
tional ndings of social support, depression and inammatory
gene expression associations, ovarian cancer patients with a great-
4
Multimorbidity is dened by the simultaneous existence of more than one
er sense of social attachment had a lower likelihood of death (Lut-
pathophysiologic condition or clinical entity (p. 371; Ritchie et al., 2011). gendorf et al., 2012). Lastly, perceived social isolation or loneliness
S4 P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9

predicts morbidity and mortality risk across different age groups integrative approach will be necessary for the next steps in re-
(Perissinotto et al., 2012; Udell et al., 2012). search that target both mechanisms and interventions.

7. Bioecological perspective of cancer and cancer survivorship 9. Overview of invited reviews and empirical reports in
Advances in Cancer and Brain, Behavior, and Immunity
These data highlight the potential utility of life course/life span
or bioecological perspectives of cancer and cancer survivorship. Scholars in PNI and related disciplines and in cancer research
Most models of mortality and survival rely on tumor characteris- were invited to author the papers contained in this volume. Reec-
tics and treatment exposure as prognostic indicators (Merletti tive of the decade that bore witness to the sequencing of the hu-
et al., 2011; Ward et al., 2004; Wei et al., 2010). Tumors develop man genome, the Cole review highlights several conceptual and
within microenvironments, yet cancers develop within a person methodological innovations that are transforming our knowledge
nested within several environmental contexts. Colditz and Wei of neural and endocrine regulation of the cancer genome (Cole,
(2012) assert that traditional projections of cancer mortality fail 2013). Sood and colleagues review studies that have converged
to account adequately for multilevel interactions and reciprocity to rene our understanding of sympathetic nervous system regula-
among biologic pathways, physical/built environment, and social/ tion of pathways relevant to cancer growth and progression (Ar-
behavioral factors (Colditz and Wei, 2012). Models that dynami- maiz-Pena et al., 2012). The nuanced and selective inuences of
cally capture exposure to multiple risk and protective factors, cer- neuroendocrine hormones on tumor cells, stromal cells and the
tain to have pleiotropic effects across and within time and levels of metastatic cascade have come into focus and are beginning to re-
analysis (Evans and Kim 2010), promise to reveal greater under- veal new therapeutic opportunities. Volden and Conzen present a
standing of individual- and population-level differences in cancer complementary review of the inuence of glucocorticoid signaling
risk and outcome (Gehlert et al., 2008; Hiatt and Breen, 2008; War- on tumor progression through cell context-specic transcriptional
necke et al., 2008). Inequalities in cancer incidence, mortality, and networks (Volden and Conzen, 2012 1045). In the clinical context,
survival by race/ethnicity and socioeconomic status prevail5 disruption of HPA rhythms, as indicated by diurnal cortisol slopes,
(Chang et al., 2012; Merletti et al., 2011; Ward et al., 2004). A grow- predicted early metastatic breast cancer mortality (Sephton et al.,
ing literature denes the biology of [social] disadvantage and early 2000). Sephton and colleagues, as reported in this volume, repli-
adversity and offers tenable hypotheses and mechanistic pathways cate those ndings in a small sample of lung cancer patients fol-
as explanations for disparities in health and disease outcomes across lowed for a median of 4 years from date of diagnosis (Sephton
the lifespan (Adler and Stewart, 2010; Boyce et al., 2012; Kelly-Irving et al., 2012). Volden and Conzen foreshadow emerging interest in
et al., 2012). We use this platform to encourage deliberate invest- stress regulation of epithelial cancer biology through metabolic
ment in research on biopsychosocial mechanisms associated with pathways and energy regulators such as insulin, leptin, ghrelin,
persistent disparities in cancer outcome (Parente et al., 2012). and adiponectin (Cao and During, 2012; Williams et al., 2009).
Convergence of animal models and human correlative studies
led Neeman and Ben-Eliyahu to identify catecholamine and prosta-
8. Animal models of human disease: powerful tools for glandin-mediated immunosuppression as a perioperative risk fac-
elucidation of mechanisms tor for cancer recurrence and metastasis (Neeman and Ben-
Eliyahu, 2012). The authors advance a theoretical model that cap-
Use of correlation studies to support weight of the evidence tures the cumulative risk and review mechanistic support for the
has been a prevalent criticism levied against PNI studies of cancer. use of pharmacological blockade of key mediators during the per-
However, within the last decade, growing availability of transgenic ioperative period. Sheridan and colleagues review the utility of a
and knockout mouse models of human cancer provides opportuni- mouse model of repeated social defeat to elucidate neural-immune
ties to understand how PNI-type interactions may modulate the mechanisms in cancer (Powell et al., 2012). This review highlights
molecular biology of cancer. Orthotopic and human tumor xeno- the role of myeloid-derived cells in stress-primed inammation, in
graft models more accurately recapitulate the dynamics of human tissue remodeling in non-immune and immune organs, and in sup-
cancer in vivo (Talmadge et al., 2007). Biologically sophisticated port of behavioral states experienced as cancer-associated sickness
animal models of human cancer provide a context for experimental behaviors (see reviews in this volume by Bower and Lamkin, 2012;
manipulation of psychosocial factors, such as environmental Costanzo et al., 2012; Irwin et al., 2012). The empirical paper by
enrichment (Cao et al., 2010), isolation (Hermes and McClintock, Madden et al. examines the impact of social isolation on breast
2008), stress (Sheridan et al., 2004; Thaker et al., 2006), and cancer pathogenesis in adult severe combined immunodeciency
depression (Lamkin et al., 2011). In addition, animal models ad- mice using a human breast cancer cell line known to express b-
vance the discovery of the consequent changes in neuronal struc- ARs (Madden et al., 2012). The results raise implications of mild
ture and function, neuroendocrine and immune activity, and vs. chronic stress exposure, timing of exposure during the life span
peripheral biology that inuence tumor cells and their microenvi- of experimental animals, and the need to capture transient shifts in
ronment. In this conceptualization, psychosocial factors set the target cell populations. Further, the study supports the importance
stage for a macroenvironment that can shape tumor microenvi- of myeloid-derived suppressor cells and stress-associated leuko-
ronments to be more or less favorable to tumor growth. This sys- cyte recruitment as indicated by changes in macrophage popula-
tems-approach highlights the interactions of networks of pro- tions in tumor and spleen, similar to that observed with social
tumor and anti-tumor mechanisms, and underscores the multiple disruption (SDR) stress paradigms (Engler et al., 2004; Powell
processes involved in both biobehavioral contributions to tumor et al., 2012).
growth, as well as in resistance to tumor growth. Such a broad, Bower and Lamkin identify two questions that direct contempo-
rary research on cancer-related fatigue, i.e., what are the neural
5
Social ecological theory predominately guides the exploration of the fundamental underpinnings of fatigue that are distinct from depression, and
causes of disparities in health. A thoughtful and balanced reection of the relative what are the factors that contribute to inammation and fatigue
contributions of local, State, and Federal level policies, social and physical environ-
ments, and access to and quality of health services is beyond the scope of this
at different treatment phases (Bower and Lamkin, 2012)? The re-
commentary and research contained in this volume. For recent reviews and view calls for established animal models of cancer-related fatigue,
perspectives see; Colditz et al., 2012 and Esnaola and Ford, 2012. attention to signal transduction pathways and downstream mark-
P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9 S5

ers of inammatory activation, prospective longitudinal studies of hippocampal volume and peripheral cytokine levels in a sample of
fatigue to map time course and recovery, and exploration of mech- breast cancer survivors nearly ve years post-chemotherapy expo-
anisms that might explain observations of symptom clusters. The sure (Kesler et al., 2012). The Kesler et al. and Ganz et al. papers re-
authors suggest early life stress as a plausible risk factor for inam- port associations between tumor necrosis factor-alpha (TNF-a) and
mation that undergirds cancer-related fatigue. The empirical paper memory impairments. McDonald and colleagues replicate and ex-
by Witek-Jansek et al. in this volume explores whether childhood tend prior work by their group and others (Kesler et al., 2011;
adversity is associated with vulnerability for intense sustained McDonald et al., 2010). Their current study reports chemother-
behavioral symptoms, including fatigue and depressive symptoms, apy-associated structural brain changes in frontal regions that cor-
and quality of life and immune dysregulation (Witek Janusek et al., respond to concurrent perceptions of compromised executive
2012). function (McDonald et al., 2012). We recognize that these studies
Irwin and colleagues describe the common presentation of sleep have limitations such as small samples sizes, discordance between
disturbance and depression in cancer survivors (Irwin et al., 2012). objective cognitive performance and subjective complaints, and, in
The authors outline a model in which sleep disturbance drives some cases, lack of pre/post-treatment and/or non-cancer control
alterations in inammatory biology, which result in of depressive comparisons. These limitations beg for prospective longitudinal
symptoms and in clinical depression for some. The model acknowl- designs that facilitate pooling of data from different research
edges depression history and other psychosocial, biobehavioral, groups, harmonization of measures, and the use of advanced statis-
and medical factors that might act as moderators. The Lutgendorf tical methods and modeling (Nelson and Suls, in press). Neverthe-
laboratory contributes an analysis of associations between cortisol, less, research presented by Ganz et al., Kesler et al., and McDonald
interleukin-6, depression, fatigue, and disability in ovarian cancer et al. nicely illustrates the nexus of brain, behavior, and
patients followed prospectively from pre-surgical baseline to inammation.
one-year post surgery, and illustrates how chemotherapy acts to
normalize these biological markers (Schrepf et al., 2012).
Although challenges exist, the review by Costanzo et al. identi- 10. Through the looking glass
es opportunities to explore clinically signicant PNI relationships
in a hematopoietic stem cell transplantation context (HSCT) (Cost- This supplement synthesizes contemporary understanding of
anzo et al., 2012). Improved understanding of the factors that mod- PNI in a cancer context and suggests opportunities for further dis-
erate timely immune recovery and optimal immune regulation covery of mechanisms and development of interventions to im-
might confer improved short- and long-term outcomes for HSCT prove clinical cancer care. Multiple signaling pathways by which
recipients. Noted as challenges for PNI researchers working in a the macroenvironment can inuence the tumor microenviron-
HSCT context are the pace of change and evolution in HSCT medi- ment are identied, but many unanswered questions remain. For
cine and associated technical innovations. The secondary data example, numerous effects of catecholaminergic and glucocorti-
analysis by McGregor et al. investigating the effect of pre-trans- coid signaling on tumor growth and progression have begun to
plantation distress on white blood cell count among autologous be mapped, but it is likely that there are multiple downstream ef-
hematopoietic cell transplantation patients, highlights these chal- fects on tumor growth processes, many of which have not been
lenges (McGregor et al., 2012). identied (for example, see Zappala et al., 2012). Hanahan and
Within the last decade, exercise has been established as an Weinberg (2011), in an update to their classic paper, highlighted
effective adjuvant therapy to control adverse consequences associ- 10 hallmarks of cancer that are necessary for tumor growth and
ated with cancer treatment. Jones et al. comprehensively reviews progression. These include sustaining proliferative signaling; evad-
extant evidence linking exercise behavior, functional capacity/ ing growth suppressors; avoiding immune destruction; enabling
exercise capacity, disease recurrence, and cancer-specic and all- replicative immortality; tumor-promoting inammation; activat-
cause mortality (Betof et al., 2012). Further, the authors outline ing invasion and metastasis, inducing angiogenesis; genome insta-
host and tumor-related mechanisms underlying the exercise/t- bility and mutation; resisting cell death; and deregulating cellular
ness and prognosis relationship and review evidence from pre-clin- energetics. The work highlighted in this issue describes how the
ical animal models of cancer. This exciting work highlights exercise stress response can inuence the macroenvironment to support
as one critical component of energy balance inuences on cancer these hallmarks.
etiology, progression, and outcome (Hursting et al., 2012). In addition to effects on the tumor and microenvironment,
This volume would not be complete without a balanced synthe- there are likely multiple upstream biobehaviorally modulated
sis of extant literature on psychological and physiological adapta- pathways that may affect tumor growth, which will make produc-
tion and psychosocial interventions following a diagnosis of tive targets for future investigation. These include the role of the
cancer (Antoni, 2012). Antoni notes the opportunity to consider parasympathetic nervous system, of biobehaviorally sensitive neu-
outcomes beyond survival and disease recurrence, the importance ropeptides and hormones such as oxytocin, prolactin, growth hor-
of determining optimal timing of interventions, acknowledgment mone, and prostaglandins, as well as a variety of metabolic
of cancers as different diseases, and the need to identify individuals mediators (e.g. insulin growth factor-1, leptin, and ghrelin) that
at high risk for poor outcomes. He discusses application of micro- are sensitive to biobehavioral pathways. Biobehavioral mediators
array and bioinformatic analyses (Cole, 2010; Cole et al., 2005) to seldom work alone, and yet mechanistic research has focused on
demonstrate that an intervention can causally inuence inamma- investigation of discrete pathways for the sake of dening mecha-
tory and metastasis-regulated gene expression in circulating leuko- nisms. However, to understand the relevant mechanisms, it will be
cytes from early-stage breast cancer patients (Antoni et al., 2012). important to understand downstream effects of interconnected
Three empirical papers in this volume focus on cognitive dys- pathways e.g., the synergistic effects on tumor dynamics of NE
function due to cancer treatment exposure in breast cancer sam- and cortisol in chronic stress. We envision a complex web of sys-
ples (Ganz et al., 2012; Kesler et al., 2012; McDonald et al., temic pathways that inuence tumor growth and development at
2012). Ganz et al. conducted an interim cross-sectional analysis multiple levels. This critical information will guide understanding
of a prospective, longitudinal, observational cohort study to ex- of whether therapies can be successful by blocking only adrenergic
plore associations between proinammatory cytokines, cerebral signaling (as in use of beta-blockers), or whether adrenergic signal-
functioning, and chemotherapy exposure (Ganz et al., 2012). Sim- ing and prostaglandins must be jointly blocked (see Neeman and
ilarly, Kesler and colleagues investigated the correlations between Ben-Eliyahu, 2012), or whether adrenergic and glucocorticoid
S6 P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9

pathways must both be targeted. Understanding whether narrow the National Cancer Institute Division of Cancer Control and Popula-
or broad targeting of therapies is clinically indicated is critical in tion Sciences. Lastly, we thank the Brain, Behavior, and Immunity se-
developing successful pharmacologic approaches. Behavioral inter- nior editorial staff for their support of this special issue.
ventions tend to be broad spectrum- targeting many overlapping
biobehavioral pathways; future research on behavioral interven- Conict of interest
tions may benet from analysis of which molecular pathways are
active. The authors of this manuscript have nothing to declare.
Future research will also benet from parsing out effects of dif-
ferent biobehavioral states e.g. stress, depression, social isolation
Acknowledgments
to determine if there is one nal common pathway, or to what
extent there are discrete biological signatures of these different
Nicole Saiontz provided editorial support and Kate McNeil pro-
psychological constructs. Molecular signatures of positive con-
vided administrative management for the special issue. The Na-
structs also need further investigation. For example, does resilience
tional Cancer Institute Network on Biobehavioral Pathways in
just mean less sympathetic activation or less hormonal and inam-
Cancer provided scientic consultation for the development of
matory responsivity to stress, or does it mean greater parasympa-
the Figure. Figure illustration by Ethan Tyler. Figure design by Will
thetic tone, or differential signaling of pathways such as those
Bramlett.
involving oxytocin or dopamine? Likewise, it is not known whether
stress factors act in a relatively linear doseresponse fashion or
References
whether there are thresholds for stress/depression/social isolation
that determine physiological trajectories that will inuence the Adler, N.E., Stewart, J., 2010. Preface to the biology of disadvantage: socioeconomic
clinical course of cancer. These kinds of data will help us better status and health. Ann. N.Y. Acad. Sci. 1186, 14.
understand who will most benet from behavioral or pharmaco- Albini, A., Sporn, M.B., 2007. The tumour microenvironment as a target for
chemoprevention. Nat. Rev. Cancer 7, 139147.
logical interventions to reduce adrenergic signaling or stress re- Andersen, B.L., Farrar, W.B., Golden-Kreutz, D., Kutz, L.A., MacCallum, R., Courtney,
sponse states - for example, what levels of stress/distress are M.E., Glaser, R., 1998. Stress and immune responses after surgical treatment for
necessary at the outset for an intervention to make a difference. regional breast cancer. J. Natl. Cancer Inst. 90, 3036.
Andersen, B.L., Kiecolt-Glaser, J.K., Glaser, R., 1994. A biobehavioral model of cancer
Moreover, the use of discrete interventions is useful for mechanis- stress and disease course. Am. Psychol. 49, 389404.
tic research purposes, but it is possible that multifaceted total life- Antoni, M.H., 2003. Psychoneuroendocrinology and psychoneuroimmunology of
style interventions that address stress factors, as well as nutritional cancer: plausible mechanisms worth pursuing? Brain Behav. Immun. 17 (Suppl.
1), S84S91.
and exercise lifestyle components, will be necessary to profoundly
Antoni, M.H., 2012. Psychosocial intervention effects on adaptation, disease course
impact cancer growth. To date, research on multimodal interven- and biobehavioral processes in cancer. Brain Behav. Immun., http://dx.doi.org/
tions remains quite limited. 10.1016/j.bbi.2012.05.009.
Additionally, the effects of biobehavioral pathways on recovery Antoni, M.H., Lechner, S., Diaz, A., Vargas, S., Holley, H., Phillips, K., McGregor, B.,
Carver, C.S., Blomberg, B., 2009. Cognitive behavioral stress management effects
from specic cancer treatments such as HSCT, adoptive immuno- on psychosocial and physiological adaptation in women undergoing treatment
therapy, surgical recovery, are important frontiers for future work. for breast cancer. Brain Behav. Immun. 23, 580591.
Understanding tumor and treatment effects on the central nervous Antoni, M.H., Lutgendorf, S.K., Blomberg, B., Carver, C.S., Lechner, S., Diaz, A., Stagl, J.,
Arevalo, J.M., Cole, S.W., 2012. Cognitive-behavioral stress management
system are equally important. As reported by some of the papers in reverses anxiety-related leukocyte transcriptional dynamics. Biol. Psychiatry
this volume, we are just beginning to understand the relevant biol- 1, 366372.
ogy in post-chemotherapy fatigue and cognitive difculties this Antoni, M.H., Lutgendorf, S.K., Cole, S.W., Dhabhar, F.S., Sephton, S.E., McDonald,
P.G., Stefanek, M., Sood, A.K., 2006. The inuence of bio-behavioural factors on
type of mechanistic understanding is critical before new treat- tumour biology: pathways and mechanisms. Nat. Rev. Cancer 6, 240248.
ments can be developed and tested. Armaiz-Pena, G.N., Cole, S.W., Lutgendorf, S.K., Sood, A.K., 2012. Neuroendocrine
Future directions also include determination of what are the inuences on cancer progression. Brain Behav. Immun., http://dx.doi.org/
10.1016/j.bbi.2012.06.005.
most important intermediate outcome variables for biobehavioral Badino, G.R., Novelli, A., Girardi, C., Di Carlo, F., 1996. Evidence for functional beta-
cancer research. In addition to overall survival and progression- adrenoceptor subtypes in CG-5 breast cancer cell. Pharmacol. Res. 33, 255260.
free survival, to what extent are gene signatures, metabolomics, Betof, A.S., Dewhirst, M.W., Jones, L.W., 2012. Effects and potential mechanisms of
exercise training on cancer progression: a translational perspective. Brain
and epigenetic changes important outcomes for this work? The re-
Behav. Immun.
search in this volume points to the dramatic discoveries that have Bovbjerg, D.H., Valdimarsdottir, H., 1993. Familial cancer, emotional distress, and
been made in the last decade to dene this eld. Future research low natural cytotoxic activity in healthy women. Ann. Oncol. 4, 745752.
holds promise for discovery of novel biobehavioral signaling path- Bower, J.E., 2012. Fatigue, brain, behavior, and immunity: summary of the 2012
Named Series on fatigue. Brain Behav. Immun. 26, 12201223.
ways that are relevant to cancer and a greater understanding of Bower, J.E., Ganz, P.A., Irwin, M.R., Kwan, L., Breen, E.C., Cole, S.W., 2011.
behavioral, pharmacologic, and complementary interventions that Inammation and behavioral symptoms after breast cancer treatment: do
target these mechanisms. fatigue, depression, and sleep disturbance share a common underlying
mechanism? J. Clin. Oncol. 29, 35173522.
In conclusion, we would be remiss if we did not thank lead Bower, J.E., Lamkin, D.M., 2012. Inammation and cancer-related fatigue:
authors and their authorship teams for contributing scientic ad- mechanisms, contributing factors, and treatment implications. Brain Behav.
vances relevant to this volume. These individuals and many others Immun., http://dx.doi.org/10.1016/j.bbi.2012.06.011.
Boyce, W.T., Sokolowski, M.B., Robinson, G.E., 2012. Toward a new biology of social
have worked quite tirelessly to improve methodological rigor, adversity. Proc. Natl. Acad. Sci. 109, 1714317148.
establish causation as appropriate, collaborate in the spirit of Braithwaite, D., Moore, D.H., Satariano, W.A., Kwan, M.L., Hiatt, R.A., Kroenke, C.,
transdisciplinary team science, and move between different re- Caan, B.J., 2012. Prognostic impact of comorbidity among long-term breast
cancer survivors: results from the LACE study. Cancer Epidemiol. Biomarkers
search designs to test and conrm experimental and clinical nd- Prev. 21, 11151125.
ings. We thank the many scholars who engaged in the peer Cao, L., During, M.J., 2012. What is the brain-cancer connection? Annu. Rev.
review process to vet the invited mini-reviews and empirical pa- Neurosci. 35, 331345.
Cao, L., Liu, X., Lin, E.J., Wang, C., Choi, E.Y., Riban, V., Lin, B., During, M.J., 2010.
pers that comprise this supplement. We acknowledge the inspira-
Environmental and genetic activation of a brain-adipocyte BDNF/leptin axis
tion and contributions of the National Cancer Institute Network on causes cancer remission and inhibition. Cell 142, 5264.
Biobehavioral Pathways in Cancer6 and the invaluable support of Castano, Z., Tracy, K., McAllister, S.S., 2011. The tumor macroenvironment and
systemic regulation of breast cancer progression. Int. J. Dev. Biol. 55, 889897.
Chang, C.M., Su, Y.C., Lai, N.S., Huang, K.Y., Chien, S.H., Chang, Y.H., Lian, W.C., Hsu,
T.W., Lee, C.C., 2012. The combined effect of individual and neighborhood
6
See http://cancercontrol.cancer.gov/brp/bbpsb/ncintwk-biopthwys.html. socioeconomic status on cancer survival rates. PLoS One 7, e44325.
P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9 S7

Chida, Y., Hamer, M., Wardle, J., Steptoe, A., 2008. Do stress-related psychosocial D.R., Lefkowitz, R.J., 2011. A stress response pathway regulates DNA damage
factors contribute to cancer incidence and survival? Nat. Clin. Pract. Oncol. 5, through beta2-adrenoreceptors and beta-arrestin-1. Nature 477, 349353.
466475. Haroon, E., Raison, C.L., Miller, A.H., 2012. Psychoneuroimmunology meets
Choi, M., Craft, B., Geraci, S.A., 2011. Surveillance and monitoring of adult cancer neuropsychopharmacology: translational implications of the impact of
survivors. Am. J. Med. 124, 598601. inammation on behavior. Neuropsychopharmacology 37, 137162.
Cohen, L., Cole, S.W., Sood, A.K., Prinsloo, S., Kirschbaum, C., Arevalo, J.M., Jennings, Hart, I.R., Fidler, I.J., 1980. Cancer invasion and metastasis. Q. Rev. Biol. 55, 121142.
N.B., Scott, S., Vence, L., Wei, Q., Kentor, D., Radvanyi, L., Tannir, N., Jonasch, E., Hermes, G.L., McClintock, M.K., 2008. Isolation and the timing of mammary gland
Tamboli, P., Pisters, L., 2012. Depressive symptoms and cortisol rhythmicity development, gonadarche, and ovarian senescence. Implications for mammary
predict survival in patients with renal cell carcinoma: role of inammatory tumor burden. Dev. Psychobiol. 50, 353360.
signaling. PLoS One 7, e42324. Hiatt, R.A., Breen, N., 2008. The social determinants of cancer: a challenge for
Cohen, S., Rabin, B.S., 1998. Psychologic stress, immunity, and cancer. J. Natl. Cancer transdisciplinary science. Am. J Prev. Med. 35, S141S150.
Inst. 90, 34. Holt-Lunstad, J., Smith, T.B., Layton, J.B., 2010. Social relationships and mortality
Colditz, G.A., Wei, E.K., 2012. Preventability of cancer: the relative contributions of risk: a meta-analytic review. PLoS Med. 7, e1000316.
biologic and social and physical environmental determinants of cancer Hursting, S.D., Digiovanni, J., Dannenberg, A.J., Azrad, M., Leroith, D., Demark-
mortality. Annu. Rev. Public Health 33, 137156. Wahnefried, W., Kakarala, M., Brodie, A., Berger, N.A., 2012. Obesity, energy
Colditz, G.A., Wolin, K.Y., Gehlert, S., 2012. Applying what we know to accelerate balance, and cancer: new opportunities for prevention. Cancer Prev. Res. 5,
cancer prevention. Sci. Transl. Med 4, 127rv4. 12601272.
Cole, S.W., 2010. Elevating the perspective on human stress genomics. Irwin, M.R., Cole, S.W., 2011. Reciprocal regulation of the neural and innate immune
Psychoneuroendocrinology 35, 955962. systems. Nat. Rev. Immunol. 11, 625632.
Cole, S.W., 2013. Nervous system regulation of the cancer genome. Brain Behav. Irwin, M.R., Olmstead, R.E., Ganz, P.A., Haque, R., 2012. Sleep disturbance,
Immun., http://dx.doi.org/10.1016/j.bbi.2012.11.008. inammation and depression risk in cancer survivors. Brain Behav. Immun,
Cole, S.W., Sood, A.K., 2012. Molecular Pathways: beta-adrenergic signaling in http://dx.doi.org/10.1016/j.bbi.2012.05.002.
cancer. Clin. Cancer Res. 18, 12011206. Joyce, J.A., 2005. Therapeutic targeting of the tumor microenvironment. Cancer Cell
Cole, S.W., Yan, W., Galic, Z., Arevalo, J., Zack, J.A., 2005. Expression-based 7, 513520.
monitoring of transcription factor activity: the TELiS database. Bioinformatics Joyce, J.A., Pollard, J.W., 2009. Microenvironmental regulation of metastasis. Nat.
21, 803810. Rev. Cancer 9, 239252.
Costanzo, E.S., Juckett, M.B., Coe, C.L., 2012. Biobehavioral inuences on recovery Karelina, K., DeVries, A.C., 2011. Modeling social inuences on human health.
following hematopoietic stem cell transplantation. Brain Behav. Immun., http:// Psychosom. Med. 73, 6774.
dx.doi.org/10.1016/j.bbi.2012.07.005. Kelley, K.W., Bluthe, R.M., Dantzer, R., Zhou, J.H., Shen, W.H., Johnson, R.W.,
Dantzer, R., Kelley, K.W., 2007. Twenty years of research on cytokine-induced Broussard, S.R., 2003. Cytokine-induced sickness behavior. Brain Behav.
sickness behavior. Brain Behav. Immun. 21, 153160. Immun.. 17 (Suppl. 1), S112S118.
Dantzer, R., Meagher, M.W., Cleeland, C.S., 2012. Translational approaches to Kelly-Irving, M., Mabile, L., Grosclaude, P., Lang, T., Delpierre, C., 2012. The
treatment-induced symptoms in cancer patients. Nat. Rev. Clin. Oncol. 9, 414 embodiment of adverse childhood experiences and cancer development:
426. potential biological mechanisms and pathways across the life course. Int. J.
Dantzer, R., OConnor, J.C., Freund, G.G., Johnson, R.W., Kelley, K.W., 2008. From Public Health.
inammation to sickness and depression: when the immune system subjugates Kesler, S., Janelsins, M., Koovakkattu, D., Palesh, O., Mustian, K., Morrow, G.,
the brain. Nat. Rev. Neurosci. 9, 4656. Dhabhar, F.S., 2012. Reduced hippocampal volume and verbal memory
Dell, H., 2006. Milestones 1: observations from a ploughman. Nat. Rev. Cancer 6, S7. performance associated with interleukin-6 and tumor necrosis factor-alpha
Dettenborn, L., James, G.D., Berge-Landry, H., Valdimarsdottir, H.B., Montgomery, levels in chemotherapy-treated breast cancer survivors. Brain Behav. Immun.,
G.H., Bovbjerg, D.H., 2005. Heightened cortisol responses to daily stress in http://dx.doi.org/10.1016/j.bbi.2012.05.017.
working women at familial risk for breast cancer. Biol. Psychol. 69, 167179. Kesler, S.R., Kent, J.S., OHara, R., 2011. Prefrontal cortex and executive function
DeVita Jr., V.T., Rosenberg, S.A., 2012. Two hundred years of cancer research. N. Engl. impairments in primary breast cancer. Arch. Neurol. 68, 14471453.
J. Med. 366, 22072214. Kiecolt-Glaser, J.K., Glaser, R., 1999. Psychoneuroimmunology and cancer: fact or
Eheman, C., Henley, S.J., Ballard-Barbash, R., Jacobs, E.J., Schymura, M.J., Noone, A.M., ction? Eur. J. Cancer 35, 16031607.
Pan, L., Anderson, R.N., Fulton, J.E., Kohler, B.A., Jemal, A., Ward, E., Plescia, M., Kiecolt-Glaser, J.K., Gouin, J.-P., Hantsoo, L., 2010. Close relationships, inammation,
Ries, L.A., Edwards, B.K., 2012. Annual Report to the Nation on the status of and health. Neurosci. Biobehav. Rev. 35, 3338.
cancer, 19752008, featuring cancers associated with excess weight and lack of Kiecolt-Glaser, J.K., McGuire, L., Robles, T.F., Glaser, R., 2002. Emotions, morbidity,
sufcient physical activity. Cancer 118, 23382366. and mortality: new perspectives from psychoneuroimmunology. Annu. Rev.
Engler, H., Bailey, M.T., Engler, A., Sheridan, J.F., 2004. Effects of repeated social Psychol. 53, 83107.
stress on leukocyte distribution in bone marrow, peripheral blood and spleen. J. Lai, P., Rabinowich, H., Crowley-Nowick, P.A., Bell, M.C., Mantovani, G., Whiteside,
Neuroimmunol. 148, 106115. T.L., 1996. Alterations in expression and function of signal-transducing proteins
Esnaola, N.F., Ford, M.E., 2012. Racial differences and disparities in cancer care and in tumor-associated T and natural killer cells in patients with ovarian
outcomes: wheres the rub? Surg. Oncol. Clin. N. Am. 21, 417437. carcinoma. Clin. Cancer Res. 2, 161173.
Evans, G.W., Kim, P., 2010. Multiple risk exposure as a potential explanatory Lamkin, D.M., Lutgendorf, S.K., Lubaroff, D., Sood, A.K., Beltz, T.G., Johnson, A.K.,
mechanism for the socioeconomic statushealth gradient. Annals of the New 2011. Cancer induces inammation and depressive-like behavior in the mouse:
York Academy of Sciences 1186, 174189. modulation by social housing. Brain Behav. Immun. 25, 555564.
Farrell, A., 2006. Milestone 7: bloodlines. Nat. Rev. Cancer 6, S11. Land, L.H., Dalton, S.O., Jensen, M.B., Ewertz, M., 2012. Impact of comorbidity on
Fidler, I.J., 2003. The pathogenesis of cancer metastasis: the seed and soil mortality: a cohort study of 62,591 Danish women diagnosed with early breast
hypothesis revisited. Nat. Rev. Cancer 3, 453458. cancer, 19902008. Breast Cancer Res. Treat. 131, 10131020.
Folkman, J., 1971. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. Langley, R.R., Fidler, I.J., 2007. Tumor cell-organ microenvironment interactions in
285, 11821186. the pathogenesis of cancer metastasis. Endocr. Rev. 28, 297321.
Ganz, P.A., Bower, J.E., Kwan, L., Castellon, S.A., Silverman, D.H., Geist, C., Breen, E.C., Levy, S., Herberman, R., Lippman, M., dAngelo, T., 1987. Correlation of stress factors
Irwin, M.R., Cole, S.W., 2012. Does tumor necrosis factor-alpha (TNF-alpha) play with sustained depression of natural killer cell activity and predicted prognosis
a role in post-chemotherapy cerebral dysfunction? Brain Behav. Immun., http:// in patients with breast cancer. J. Clin. Oncol. 5, 348353.
dx.doi.org/10.1016/j.bbi.2012.07.015. Levy, S.M., Herberman, R.B., Maluish, A.M., Schlien, B., Lippman, M., 1985.
Ganzel, B.L., Morris, P.A., Wethington, E., 2010. Allostasis and the human brain: Prognostic risk assessment in primary breast cancer by behavioral and
integrating models of stress from the social and life sciences. Psychol. Rev. 117, immunological parameters. Health Psychol. 4, 99113.
134174. Levy, S.M., Herberman, R.B., Whiteside, T., Sanzo, K., Lee, J., Kirkwood, J., 1990.
Gehlert, S., Sohmer, D., Sacks, T., Mininger, C., McClintock, M., Olopade, O., 2008. Perceived social support and tumor estrogen/progesterone receptor status as
Targeting health disparities: a model linking upstream determinants to predictors of natural killer cell activity in breast cancer patients. Psychosom.
downstream interventions. Health Aff. 27, 339349. Med. 52, 7385.
Giese-Davis, J., Collie, K., Rancourt, K.M., Neri, E., Kraemer, H.C., Spiegel, D., 2011. Lipshultz, S.E., Landy, D.C., Lopez-Mitnik, G., Lipsitz, S.R., Hinkle, A.S., Constine, L.S.,
Decrease in depression symptoms is associated with longer survival in patients French, C.A., Rovitelli, A.M., Proukou, C., Adams, M.J., Miller, T.L., 2012.
with metastatic breast cancer: a secondary analysis. J. Clin. Oncol. 29, 413420. Cardiovascular status of childhood cancer survivors exposed and unexposed
Glaser, R., Kiecolt-Glaser, J.K., 2005. Stress-induced immune dysfunction: to cardiotoxic therapy. J. Clin. Oncol. 30, 10501057.
implications for health. Nat. Rev. Immunol. 5, 243251. Lotzova, E., Savary, C.A., Freedman, R., Bowen, J.M., 1986. Natural immunity against
Gold, S.M., Zakowski, S.G., Valdimarsdottir, H.B., Bovbjerg, D.H., 2003. Stronger ovarian tumors. Comp. Immunol. Microbiol. Infect. Dis. 9, 269275.
endocrine responses after brief psychological stress in women at familial risk of Lotzova, E., Savary, C.A., Freedman, R.S., Bowen, J.M., 1984. Natural killer cell
breast cancer. Psychoneuroendocrinology 28, 584593. cytotoxic potential of patients with ovarian carcinoma and its modulation with
Gonda, T.A., Tu, S., Wang, T.C., 2009. Chronic inammation, the tumor virus-modied tumor cell extract. Cancer Immunol. Immunother. 17, 124129.
microenvironment and carcinogenesis. Cell Cycle 8, 20052013. Lutgendorf, S.K., Cole, S., Costanzo, E., Bradley, S., Cofn, J., Jabbari, S., Rainwater, K.,
Hanahan, D., Weinberg, R.A., 2000. The hallmarks of cancer. Cell 100, 5770. Ritchie, J.M., Yang, M., Sood, A.K., 2003. Stress-related mediators stimulate
Hanahan, D., Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell vascular endothelial growth factor secretion by two ovarian cancer cell lines.
144, 646674. Clin. Cancer Res. 9, 45144521.
Hara, M.R., Kovacs, J.J., Whalen, E.J., Rajagopal, S., Strachan, R.T., Grant, W., Towers, Lutgendorf, S.K., De Geest, K., Bender, D., Ahmed, A., Goodheart, M.J., Dahmoush, L.,
A.J., Williams, B., Lam, C.M., Xiao, K., Shenoy, S.K., Gregory, S.G., Ahn, S., Duckett, Zimmerman, M.B., Penedo, F.J., Lucci, J.A., Ganjei-Azar, P., Thaker, P.H., Mendez,
S8 P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9

L., Lubaroff, D.M., Slavich, G.M., Cole, S.W., Sood, A.K., 2012. Social inuences on Pitroda, S.P., Zhou, T., Sweis, R.F., Filippo, M., Labay, E., Beckett, M.A., Mauceri, H.J.,
clinical outcomes of patients with ovarian cancer. J. Clin. Oncol. 30, 28852890. Liang, H., Darga, T.E., Perakis, S., Khan, S.A., Sutton, H.G., Zhang, W., Khodarev,
Lutgendorf, S.K., DeGeest, K., Sung, C.Y., Arevalo, J.M., Penedo, F., Lucci 3rd, J., N.N., Garcia, J.G., Weichselbaum, R.R., 2012. Tumor endothelial inammation
Goodheart, M., Lubaroff, D., Farley, D.M., Sood, A.K., Cole, S.W., 2009. Depression, predicts clinical outcome in diverse human cancers. PLoS One 7, e46104.
social support, and beta-adrenergic transcription control in human ovarian Powell, N.D., Tarr, A.J., Sheridan, J.F., 2012. Psychosocial stress and inammation in
cancer. Brain Behav. Immun. 23, 176183. cancer. Brain Behav. Immun., http://dx.doi.org/10.1016/j.bbi.2012.06.015.
Lutgendorf, S.K., Sood, A.K., 2011. Biobehavioral factors and cancer progression: Ritchie, C.S., Kvale, E., Fisch, M.J., 2011. Multimorbidity: an issue of growing
physiological pathways and mechanisms. Psychosom. Med. 73, 724730. importance for oncologists. J. Oncol. Pract. 7, 371374.
Lutgendorf, S.K., Sood, A.K., Anderson, B., McGinn, S., Maiseri, H., Dao, M., Sorosky, Satin, J.R., Linden, W., Phillips, M.J., 2009. Depression as a predictor of disease
J.I., DeGeest, K., Ritchie, J., Lubaroff, D.M., 2005. Social support, distress, and progression and mortality in cancer patients: a meta-analysis. Cancer 115,
natural killer cell activity in ovarian cancer patients. J. Clin. Oncol. 23, 7106 53495361.
7113. Schairer, C., Mink, P.J., Carroll, L., Devesa, S.S., 2004. Probabilities of death from
Madden, K.S., Szpunar, M.J., Brown, E.B., 2012. Early impact of social isolation and breast cancer and other causes among female breast cancer patients. J. Natl.
breast tumor progression in mice. Brain Behav. Immun., http://dx.doi.org/ Cancer Inst. 96, 13111321.
10.1016/j.bbi.2012.05.003. Schrepf, A., Clevenger, L., Christensen, D., Degeest, K., Bender, D., Ahmed, A.,
Mantovani, A., 2009. Cancer: inaming metastasis. Nature 457, 3637. Goodheart, M.J., Dahmoush, L., Penedo, F., Lucci 3rd, J.A., Ganjei-Azar, P.,
Mantovani, A., Allavena, P., Sica, A., Balkwill, F., 2008. Cancer-related inammation. Mendez, L., Markon, K., Lubaroff, D.M., Thaker, P.H., Slavich, G.M., Sood, A.K.,
Nature 454, 436444. Lutgendorf, S.K., 2012. Cortisol and inammatory processes in ovarian cancer
Mariotto, A.B., Rowland, J.H., Ries, L.A., Scoppa, S., Feuer, E.J., 2007. Multiple cancer patients following primary treatment: relationships with depression, fatigue,
prevalence. A growing challenge in long-term survivorship. Cancer Epidemiol. and disability. Brain Behav. Immun., http://dx.doi.org/10.1016/
Biomarkers Prev. 16, 566571. j.bbi.2012.07.022.
Marx, J., 2008. Cancer biology. All in the stroma: cancers Cosa Nostra. Science 320, Schuldt, A., 2006. Milestone 13: environmental awareness. Nat. Rev. Cancer 6, S15.
3841. Schuller, H.M., 2008. Neurotransmission and cancer: implications for prevention
McAllister, S.S., Weinberg, R.A., 2010. Tumor-host interactions: a far-reaching and therapy. Anticancer Drugs 19, 655671.
relationship. J. Clin. Oncol. 28, 40224028. Schuller, H.M., Al-Wadei, H.A.N., 2010. Neurotransmitter receptors as central
McCawley, L.J., Matrisian, L.M., 2001. Tumor progression: dening the soil round the regulators of pancreatic cancer. Future Oncol. 6, 221228.
tumor seed. Curr. Biol. 11, R25R27. Sephton, S.E., Dhabhar, F.S., Keuroghlian, A.S., Giese-Davis, J., McEwen, B.S., Ionan,
McDonald, B.C., Conroy, S.K., Ahles, T.A., West, J.D., Saykin, A.J., 2010. Gray matter A.C., Spiegel, D., 2009. Depression, cortisol, and suppressed cell-mediated
reduction associated with systemic chemotherapy for breast cancer: a immunity in metastatic breast cancer. Brain Behav. Immun. 23, 11481155.
prospective MRI study. Breast Cancer Res. Treat. 123, 819828. Sephton, S.E., Lush, E., Dedert, E.A., Floyd, A.R., Rebholz, W.N., Dhabhar, F.S., Spiegel,
McDonald, B.C., Conroy, S.K., Smith, D.J., West, J.D., Saykin, A.J., 2012. Frontal gray D., Salmon, P., 2012. Diurnal cortisol rhythm as a predictor of lung cancer
matter reduction after breast cancer chemotherapy and association with survival. Brain Behav. Immun.
executive symptoms: a replication and extension study. Brain Behav. Immun. Sephton, S.E., Sapolsky, R.M., Kraemer, H.C., Spiegel, D., 2000. Diurnal cortisol
http://dx.doi.org/10.1016/j.bbi.2012.05.007. rhythm as a predictor of breast cancer survival. J. Natl. Cancer Inst. 92, 994
McGregor, B.A., Syrjala, K.L., Dolan, E.D., Langer, S.L., Redman, M., 2012. The effect of 1000.
pre-transplant distress on immune reconstitution among adult autologous Sheridan, J.F., Padgett, D.A., Avitsur, R., Marucha, P.T., 2004. Experimental models of
hematopoietic cell transplantation patients. Brain Behav. Immun., http:// stress and wound healing. World J. Surg. 28, 327330.
dx.doi.org/10.1016/j.bbi.2012.07.020. Siegel, R., DeSantis, C., Virgo, K., Stein, K., Mariotto, A., Smith, T., Cooper, D., Gansler,
Medrek, C., Ponten, F., Jirstrom, K., Leandersson, K., 2012. The presence of tumor T., Lerro, C., Fedewa, S., Lin, C., Leach, C., Cannady, R.S., Cho, H., Scoppa, S.,
associated macrophages in tumor stroma as a prognostic marker for breast Hachey, M., Kirch, R., Jemal, A., Ward, E., 2012a. Cancer treatment and
cancer patients. BMC Cancer 12, 306. survivorship statistics, 2012. CA Cancer J. Clin. 62, 220241.
Mell, L.K., Jeong, J.H., Nichols, M.A., Polite, B.N., Weichselbaum, R.R., Chmura, S.J., Siegel, R., Naishadham, D., Jemal, A., 2012b. Cancer statistics, 2012. CA Cancer J. Clin.
2010. Predictors of competing mortality in early breast cancer. Cancer 116, 62, 1029.
53655373. Solinas, G., Germano, G., Mantovani, A., Allavena, P., 2009. Tumor-associated
Merletti, F., Galassi, C., Spadea, T., 2011. The socioeconomic determinants of cancer. macrophages (TAM) as major players of the cancer-related inammation. J.
Environ. Health 10, S7. Leukoc. Biol. 86, 10651073.
Mertens, A.C., Liu, Q., Neglia, J.P., Wasilewski, K., Leisenring, W., Armstrong, G.T., Sullivan, M.D., OConnor, P., Feeney, P., Hire, D., Simmons, D.L., Raisch, D.W., Fine,
Robison, L.L., Yasui, Y., 2008. Cause-specic late mortality among 5-year L.J., Narayan, K.M., Ali, M.K., Katon, W.J., 2012. Depression predicts all-cause
survivors of childhood cancer: the childhood cancer survivor study. J. Natl. mortality: epidemiological evaluation from the ACCORD HRQL substudy.
Cancer Inst. 100, 13681379. Diabetes Care 35, 17081715.
Miller, A.H., Ancoli-Israel, S., Bower, J.E., Capuron, L., Irwin, M.R., 2008. Talmadge, J.E., Singh, R.K., Fidler, I.J., Raz, A., 2007. Murine models to evaluate novel
Neuroendocrine-immune mechanisms of behavioral comorbidities in patients and conventional therapeutic strategies for cancer. Am. J. Pathol. 170, 793804.
with cancer. J. Clin. Oncol. 26, 971982. Thaker, P.H., Han, L.Y., Kamat, A.A., Arevalo, J.M., Takahashi, R., Lu, C., Jennings, N.B.,
Nathan, C., 2002. Points of control in inammation. Nature 420, 846852. Armaiz-Pena, G., Bankson, J.A., Ravoori, M., Merritt, W.M., Lin, Y.G., Mangala,
Neeman, E., Ben-Eliyahu, S., 2012. Surgery and stress promote cancer metastasis: L.S., Kim, T.J., Coleman, R.L., Landen, C.N., Li, Y., Felix, E., Sanguino, A.M.,
new outlooks on perioperative mediating mechanisms and immune Newman, R.A., Lloyd, M., Gershenson, D.M., Kundra, V., Lopez-Berestein, G.,
involvement. Brain Behav. Immun., http://dx.doi.org/10.1016/j.bbi.2012.03.006. Lutgendorf, S.K., Cole, S.W., Sood, A.K., 2006. Chronic stress promotes tumor
Nelson, E.L., Wenzel, L.B., Osann, K., Dogan-Ates, A., Chantana, N., Reina-Patton, A., growth and angiogenesis in a mouse model of ovarian carcinoma. Nat. Med. 12,
Laust, A.K., Nishimoto, K.P., Chicz-DeMet, A., du Pont, N., Monk, B.J., 2008. Stress, 939944.
immunity, and cervical cancer: biobehavioral outcomes of a randomized Thornton, L.M., Andersen, B.L., Crespin, T.R., Carson, W.E., 2007. Individual
clinical trial [corrected]. Clin. Cancer Res. 14, 21112118. trajectories in stress covary with immunity during recovery from cancer
Nelson, W., Suls, J., in press. New approaches to study cognitive changes associated diagnosis and treatments. Brain Behav. Immun. 21, 185194.
with chemotherapy for non-central nervous system tumors. J. Pain Symptom Udell, J.A., Steg, P.G., Scirica, B.M., Smith Jr., S.C., Ohman, E.M., Eagle, K.A., Goto, S.,
Manage. Cho, J.I., Bhatt, D.L., 2012. Living alone and cardiovascular risk in outpatients at
Oefnger, K.C., Mertens, A.C., Sklar, C.A., Kawashima, T., Hudson, M.M., Meadows, risk of or with atherothrombosis. Arch. Intern. Med. 172, 10861095.
A.T., Friedman, D.L., Marina, N., Hobbie, W., Kadan-Lottick, N.S., Schwartz, C.L., Valdivieso, M., Kujawa, A.M., Jones, T., Baker, L.H., 2012. Cancer survivors in the
Leisenring, W., Robison, L.L., 2006. Chronic health conditions in adult survivors United States: a review of the literature and a call to action. Int. J. Med. Sci. 9,
of childhood cancer. N. Engl. J. Med. 355, 15721582. 163173.
Oefnger, K.C., Tonorezos, E.S., 2011. The cancer is over, now what?: understanding Vogelstein, B., Kinzler, K.W., 2004. Cancer genes and the pathways they control. Nat.
risk, changing outcomes. Cancer 117, 22502257. Med. 10, 789799.
Paget, S., 1889. The distribution of secondary growths in cancer of the breast. Lancet Volden, P.A., Conzen, S.D., 2012. The inuence of glucocorticoid signaling on tumor
1, 571573. progression. Brain Behav. Immun., http://dx.doi.org/10.1016/j.bbi.2012.10.022.
Parente, V., Hale, L., Palermo, T., 2012. Association between breast cancer and Ward, E., Jemal, A., Cokkinides, V., Singh, G.K., Cardinez, C., Ghafoor, A., Thun, M.,
allostatic load by race. national health and nutrition examination survey 1999- 2004. Cancer disparities by race/ethnicity and socioeconomic status. CA Cancer
2008. Psychooncology. J. Clin. 54, 7893.
Parry, C., Kent, E.E., Mariotto, A.B., Alfano, C.M., Rowland, J.H., 2011. Cancer Warnecke, R.B., Oh, A., Breen, N., Gehlert, S., Paskett, E., Tucker, K.L., Lurie, N.,
survivors: a booming population. Cancer Epidemiol. Biomarkers Prev. 20, 1996 Rebbeck, T., Goodwin, J., Flack, J., Srinivasan, S., Kerner, J., Heurtin-Roberts, S.,
2005. Abeles, R., Tyson, F.L., Patmios, G., Hiatt, R.A., 2008. Approaching health
Patnaik, J.L., Byers, T., Diguiseppi, C., Denberg, T.D., Dabelea, D., 2011. The inuence disparities from a population perspective: the National Institutes of Health
of comorbidities on overall survival among older women diagnosed with breast Centers for Population Health and Health Disparities. Am. J. Public Health 98,
cancer. J. Natl. Cancer Inst. 103, 11011111. 16081615.
Perissinotto, C.M., Stijacic Cenzer, I., Covinsky, K.E., 2012. Loneliness in older Webster Marketon, J.I., Glaser, R., 2008. Stress hormones and immune function. Cell
persons: a predictor of functional decline and death. Arch. Intern. Med. 172, Immunol. 252, 1626.
10781083. Wei, E.K., Wolin, K.Y., Colditz, G.A., 2010. Time course of risk factors in cancer
Pinquart, M., Duberstein, P.R., 2010. Depression and cancer mortality: a meta- etiology and progression. J. Clin. Oncol. 28, 40524057.
analysis. Psychol. Med. 40, 17971810.
P. Green McDonald et al. / Brain, Behavior, and Immunity 30 (2013) S1S9 S9

Williams, J.B., Pang, D., Delgado, B., Kocherginsky, M., Tretiakova, M., Krausz, T., associated with inhibition of apoptosis in breast epithelial cells. Cancer Res. 64,
Pan, D., He, J., McClintock, M.K., Conzen, S.D., 2009. A model of gene- 17571764.
environment interaction reveals altered mammary gland gene expression and Wyman, L., Crum, R.M., Celentano, D., 2012. Depressed mood and cause-specic
increased tumor growth following social isolation. Cancer Prev. Res. 2, 850 mortality: a 40-year general community assessment. Ann. Epidemiol. 22, 638
861. 643.
Williams, R.B., 2008. Psychosocial and biobehavioral factors and their interplay in Yood, M.U., Wells, K.E., Alford, S.H., Dakki, H., Beiderbeck, A.B., Hurria, A., Gross, C.P.,
coronary heart disease. Annu. Rev. Clin. Psychol. 4, 349365. Oliveria, S.A., 2012. Cardiovascular outcomes in women with advanced breast
Witek Janusek, L., Tell, D., Albuquerque, K., Mathews, H.L., 2012. Childhood cancer exposed to chemotherapy. Pharmacoepidemiol. Drug Saf. 21, 818827.
adversity increases vulnerability for behavioral symptoms and immune Zappala, G., McDonald, P.G., Cole, S.W., 2012. Tumor dormancy and the
dysregulation in women with breast cancer. Brain Behav. Immun., http:// neuroendocrine system: an undisclosed connection? Cancer Metastasis Rev.
dx.doi.org/10.1016/j.bbi.2012.05.014. Zucca, A.C., Boyes, A.W., Linden, W., Girgis, A., 2012. Alls well that ends well?
Wu, W., Chaudhuri, S., Brickley, D.R., Pang, D., Karrison, T., Conzen, S.D., 2004. Quality of life and physical symptom clusters in long-term cancer survivors
Microarray analysis reveals glucocorticoid-regulated survival genes that are across cancer types. J. Pain Symptom Manage. 43, 720731.

S-ar putea să vă placă și